1
|
SOD2, a Potential Transcriptional Target Underpinning CD44-Promoted Breast Cancer Progression. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030811. [PMID: 35164076 PMCID: PMC8839817 DOI: 10.3390/molecules27030811] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 11/17/2022]
Abstract
CD44, a cell-adhesion molecule has a dual role in tumor growth and progression; it acts as a tumor suppressor as well as a tumor promoter. In our previous work, we developed a tetracycline-off regulated expression of CD44's gene in the breast cancer (BC) cell line MCF-7 (B5 clone). Using cDNA oligo gene expression microarray, we identified SOD2 (superoxide dismutase 2) as a potential CD44-downstream transcriptional target involved in BC metastasis. SOD2 gene belongs to the family of iron/manganese superoxide dismutase family and encodes a mitochondrial protein. SOD2 plays a role in cell proliferation and cell invasion via activation of different signaling pathways regulating angiogenic abilities of breast tumor cells. This review will focus on the findings supporting the underlying mechanisms associated with the oncogenic potential of SOD2 in the onset and progression of cancer, especially in BC and the potential clinical relevance of its various inhibitors.
Collapse
|
2
|
Hu HJ, Liang X, Li HL, Wang HY, Gu JF, Sun LY, Xiao J, Hu JQ, Ni AM, Liu XY. Optimization of the Administration Strategy for the Armed Oncolytic Adenovirus ZD55-IL-24 in Both Immunocompromised and Immunocompetent Mouse Models. Hum Gene Ther 2021; 32:1481-1494. [PMID: 34155929 DOI: 10.1089/hum.2021.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
ZD55-IL-24 is an armed oncolytic adenovirus similar but superior to ONYX-015. Virotherapeutic strategies using ZD55-IL-24 have been demonstrated to be effective against several cancer types. However, it is unclear whether the traditional administration strategy is able to exert the maximal antitumor efficacy of ZD55-IL-24. In this study, we sought to optimize the administration strategy of ZD55-IL-24 in both A375-bearing immunocompromised mouse model and B16-bearing immunocompetent mouse model. Although the underlying antitumor mechanisms are quite different, the obtained results are similar in these two mouse tumor models. We find that the antitumor efficacy of ZD55-IL-24 increases as injection times increase in both of these two models. However, no obvious increase of efficacy is observed as the dose of each injection increases. Our further investigation reveals that the administration strategy of sustained ZD55-IL-24 therapy can achieve a better therapeutic effect than the traditional administration strategy of short-term ZD55-IL-24 therapy. Furthermore, there is no need to inject every day; every 2 or 3 days of injection achieves an equivalent therapeutic efficacy. Finally, we find that the sustained rather than the traditional short-term ZD55-IL-24 therapy can synergize with anti-PD-1 therapy to reject tumors in B16-bearing immunocompetent mouse model. These findings suggest that the past administration strategy of ZD55-IL-24 is in fact suboptimal and the antitumor efficacy can be further enhanced through administration strategy optimization. This study might shed some light on the development of clinically applicable administration regimens for ZD55-IL-24 therapy.
Collapse
Affiliation(s)
- Hai-Jun Hu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiu Liang
- School of Life Sciences and Technology, Tongji University, Shanghai, China; and
| | - Hai-Lang Li
- Department of Pharmacy, Xiamen Medical College, Xiamen, China
| | - Huai-Yuan Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jin-Fa Gu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Lan-Ying Sun
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Jing Xiao
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jin-Qing Hu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Ai-Min Ni
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Xin-Yuan Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
3
|
El-Mahdy MA, Alzarie YA, Hemann C, Badary OA, Nofal S, Zweier JL. The novel SOD mimetic GC4419 increases cancer cell killing with sensitization to ionizing radiation while protecting normal cells. Free Radic Biol Med 2020; 160:630-642. [PMID: 32739595 PMCID: PMC7704930 DOI: 10.1016/j.freeradbiomed.2020.07.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 07/23/2020] [Accepted: 07/25/2020] [Indexed: 02/08/2023]
Abstract
While radiotherapy is a widely used treatment for many types of human cancer, problems of radio-resistance and side effects remain. Side effects induced by ionizing radiation (IR) arise primarily from its propensity to trigger inflammation and oxidative stress with damage of normal cells and tissues near the treatment area. The highly potent superoxide dismutase mimetic, GC4419 (Galera Therapeutics), rapidly enters cells and is highly effective in dismutating superoxide (O2•-). We performed studies to assess the potency of GC4419 in cancer killing and radio-sensitization in human lung cancer cells and normal immortalized lung cells. Treatment with GC4419 did not alter the radical generation during IR, primarily hydroxyl radical (.OH); however, it quenched the increased levels of O2•- detected in the cancer cells before and following IR. GC4419 triggered cancer cell death and inhibited cancer cell proliferation with no adverse effect on normal cells. Combination of GC4419 with IR augmented the cytotoxic effects of IR on cancer cells compared to monotherapy, while protecting normal cells from IR-induced cell death. DNA fragmentation and caspase-3 activity assays showed that combination of GC4419 with IR enhances cancer cell apoptosis. Moreover, GC4419 increased IR-induced Bax levels with decreased Bcl-2 and elevated Bax/Bcl-2 ratio following treatment. GC4419 increased TrxR activity in the normal cells but decreased activity in cancer cells, conferring increased cancer cell sensitivity to oxidative stress. In conclusion, GC4419 increases the cytotoxic and pro-apoptotic activity of IR in lung cancer cells while decreasing injury in normal cells.
Collapse
Affiliation(s)
- Mohamed A El-Mahdy
- Department of Internal Medicine, Davis Heart & Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Yasmin A Alzarie
- Department of Internal Medicine, Davis Heart & Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA; National Organization of Drug Control and Research, Cairo, Egypt
| | - Craig Hemann
- Department of Internal Medicine, Davis Heart & Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Osama A Badary
- Department of Clinical Pharmacy, College of Pharmacy, The British University in Egypt, Cairo, Egypt
| | - Shahira Nofal
- Department of Pharmacology and Toxicology, College of Pharmacy, Helwan University, Cairo, Egypt
| | - Jay L Zweier
- Department of Internal Medicine, Davis Heart & Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
4
|
Xiao B, Ying C, Chen Y, Huang F, Wang B, Fang H, Guo W, Liu T, Zhou X, Huang B, Liu X, Wang Y. Doxorubicin hydrochloride enhanced antitumour effect of CEA-regulated oncolytic virotherapy in live cancer cells and a mouse model. J Cell Mol Med 2020; 24:13431-13439. [PMID: 33251723 PMCID: PMC7701578 DOI: 10.1111/jcmm.15966] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 09/19/2020] [Accepted: 09/22/2020] [Indexed: 12/12/2022] Open
Abstract
Oncolytic adenovirus (OA) has attracted increasing attention due to their specific proliferation in tumour cells and resulting in lysis of tumour cells. To further improve the antitumour effect of OA, in this study, we combined CD55-TRAIL-IETD-MnSOD (CD55-TMn), a CEA-controlled OA constructed previously, and chemotherapy to investigate their synergistic effect and possible mechanisms. MTT assay was performed to detect antitumour effects. Hoechst 33 342 and flow cytometric analysis were used to examine cell apoptosis. Western blotting was performed to examine cell pyroptosis and apoptosis mechanism. Animal experiment was used to detect antitumour effect of doxorubicin hydrochloride (Dox) combined with CD55-TMn in vivo. We firstly found that Dox promotes gene expression mediated by CEA-regulated OA and virus progeny replication by activating phosphorylation of Smad3, and Dox can enhance antitumour effect of CEA-regulated CD55-TMn by promoting cell apotopsis and cell pyroptosis. Thus, our results provide an experimental and theoretical basis on tumour therapy by combination treatment of the oncolytic virotherapy and chemotherapy and it is expected to become a novel strategy for liver cancer therapy.
Collapse
Affiliation(s)
- Boduan Xiao
- Xinyuan Institute of Medicine and BiotechnologySchool of Life Sciences and MedicineZhejiang Sci‐Tech UniversityHangzhouChina
| | - Chang Ying
- Xinyuan Institute of Medicine and BiotechnologySchool of Life Sciences and MedicineZhejiang Sci‐Tech UniversityHangzhouChina
| | - Yongyi Chen
- Institute of cancer research and basic medical sciences of Chinese Academy of SciencesCancer hospital of University of Chinese Academy of SciencesZhejiang cancer hospitalHangzhouChina
| | - Fang Huang
- Department of PathologyZhejiang Provincial People’s HospitalHangzhouChina
| | - Binrong Wang
- Xinyuan Institute of Medicine and BiotechnologySchool of Life Sciences and MedicineZhejiang Sci‐Tech UniversityHangzhouChina
| | - Huiling Fang
- Xinyuan Institute of Medicine and BiotechnologySchool of Life Sciences and MedicineZhejiang Sci‐Tech UniversityHangzhouChina
| | - Wan Guo
- Xinyuan Institute of Medicine and BiotechnologySchool of Life Sciences and MedicineZhejiang Sci‐Tech UniversityHangzhouChina
| | - Tao Liu
- Department of OtolaryngologyGuangdong General HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| | - Xiumei Zhou
- Xinyuan Institute of Medicine and BiotechnologySchool of Life Sciences and MedicineZhejiang Sci‐Tech UniversityHangzhouChina
| | - Biao Huang
- Xinyuan Institute of Medicine and BiotechnologySchool of Life Sciences and MedicineZhejiang Sci‐Tech UniversityHangzhouChina
| | - Xinyuan Liu
- Xinyuan Institute of Medicine and BiotechnologySchool of Life Sciences and MedicineZhejiang Sci‐Tech UniversityHangzhouChina
| | - Yigang Wang
- Xinyuan Institute of Medicine and BiotechnologySchool of Life Sciences and MedicineZhejiang Sci‐Tech UniversityHangzhouChina
| |
Collapse
|
5
|
Lee CL, Veeramani S, Molouki A, Lim SHE, Thomas W, Chia SL, Yusoff K. Virotherapy: Current Trends and Future Prospects for Treatment of Colon and Rectal Malignancies. Cancer Invest 2019; 37:393-414. [PMID: 31502477 DOI: 10.1080/07357907.2019.1660887] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Colorectal cancer (CRC) is one of the most common malignancies. In recent decades, early diagnosis and conventional therapies have resulted in a significant reduction in mortality. However, late stage metastatic disease still has very limited effective treatment options. There is a growing interest in using viruses to help target therapies to tumour sites. In recent years the evolution of immunotherapy has emphasised the importance of directing the immune system to eliminate tumour cells; we aim to give a state-of-the-art over-view of the diverse viruses that have been investigated as potential oncolytic agents for the treatment of CRC.
Collapse
Affiliation(s)
- Chin Liang Lee
- Perdana University-Royal College of Surgeons in Ireland School of Medicine (PU-RCSI) , Serdang , Malaysia
| | - Sanggeetha Veeramani
- Perdana University-Royal College of Surgeons in Ireland School of Medicine (PU-RCSI) , Serdang , Malaysia
| | - Aidin Molouki
- Department of Avian Disease Research and Diagnostics, Razi Vaccine and Serum Research Institute, Agricultural Research Education and Extension Organization (AREEO) , Karaj , Iran
| | - Swee Hua Erin Lim
- Perdana University-Royal College of Surgeons in Ireland School of Medicine (PU-RCSI) , Serdang , Malaysia.,Health Sciences Division, Abu Dhabi Women's College, Higher Colleges of Technology , Abu Dhabi , United Arab Emirates
| | - Warren Thomas
- Perdana University-Royal College of Surgeons in Ireland School of Medicine (PU-RCSI) , Serdang , Malaysia
| | - Suet Lin Chia
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universit Putra Malaysia , Serdang , Malaysia.,Institute of Bioscience, Universiti Putra Malaysia , Serdang , Malaysia
| | - Khatijah Yusoff
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universit Putra Malaysia , Serdang , Malaysia.,Institute of Bioscience, Universiti Putra Malaysia , Serdang , Malaysia
| |
Collapse
|
6
|
Cao X, Liu L, Yuan Q, Li X, Cui Y, Ren K, Zou C, Chen A, Xu C, Qiu Y, Quan M, Zhang J, Cao J, Chen X. Isovitexin reduces carcinogenicity and stemness in hepatic carcinoma stem-like cells by modulating MnSOD and FoxM1. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:264. [PMID: 31208440 PMCID: PMC6580799 DOI: 10.1186/s13046-019-1244-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 05/23/2019] [Indexed: 02/07/2023]
Abstract
Background Manganese superoxide dismutase (MnSOD) upregulating FoxM1 have previously been demonstrated promoting lung cancer stemness. Isovitexin exhibits antitumor activities in various cancers. This study aimed to assess whether isovitexin inhibits hepatic carcinoma stem-like cells (HCSLCs) features via regulating MnSOD and FoxM1 expression. Methods Second-generation spheres from the hepatic carcinoma cell lines, respectively, were used as HCSLCs. Protein amounts of MnSOD, FoxM1 and stemness-associated markers (CD133, CD44, ALDH1, Bmi1, Nanog and Oct4) were determined by immunoblotting. In vitro carcinogenicity was evaluated by sphere- and colony-formation assays. The effects of isovitexin on HCSLC carcinogenicity and stemness were examined in vitro and in xenograft models. An adenoviral delivery system was employed to manipulate MnSOD and/or FoxM1. Luciferase reporter assay was performed to verify isovitexin downregulated FoxM1 by inhibiting MnSOD-mediated effects of E2F1 and/or Sp1 on activation of FoxM1 promoter. Results FoxM1 upregulation by MnSOD contributed to carcinogenicity and stemness, with increased sphere- and colony-formation capabilities, upregulated stemness-associated markers and CD133+ subpopulation as well as elevated oncogenicity in vivo in HCSLCs compared with hepatic carcinoma cells. Isovitexin substantially decreased sphere and colony formation rates, and stemness-associated markers in cultured HCSLCs by suppressing MnSOD and FoxM1 expression. Importantly, isovitexin significantly inhibited tumor growth of in nude mice bearing HCSLCs and reduced CD133 protein expression of xenograft in nude mice. MnSOD or FoxM1 knockdown enhanced the effects of isovitexin suppression on carcinogenicity and stemness in HCSLC. MnSOD or FoxM1 overexpression attenuated the effects of isovitexin. Additionally, isovitexin and MnSOD knockdown could inhibit FoxM1 reporter activity via a decreased binding of E2F1 and/or Sp1 onto FoxM1 promoter. FoxM1 overexpression reversed the effects of isovitexin combined with MnSOD knockdown, without affecting MnSOD expression. Moreover, MnSOD knockdown plus thiostrepton, a FoxM1 specific inhibitor, cooperated with isovitexin to repress xenograft tumor growth and downregulate MnSOD and FoxM1 in nude mice bearing HCSLCs from MHCC97H cells. Conclusions Isovitexin inhibits carcinogenicity and stemness in HCSLCs by downregulating FoxM1via inhibition of MnSOD. Electronic supplementary material The online version of this article (10.1186/s13046-019-1244-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaocheng Cao
- Department of Pharmaceutical Science, Medical College, Hunan Normal University, Changsha, 410013, Hunan, China.,Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Changsha, 410013, Hunan, China.,Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Lihua Liu
- Pharmacy Department, the Second Clinical Medical School of Jinan University, Shenzhen People's Hospital, Shenzhen, 518020, China.,Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, Shenzhen People's Hospital, Shenzhen, 518020, China
| | - Qing Yuan
- Department of Preclinical Medicine, Medical College, Hunan Normal University, Changsha, 410013, Hunan, China
| | - Xiang Li
- Department of Preclinical Medicine, Medical College, Hunan Normal University, Changsha, 410013, Hunan, China
| | - Yinghong Cui
- Department of Pharmaceutical Science, Medical College, Hunan Normal University, Changsha, 410013, Hunan, China.,Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Changsha, 410013, Hunan, China
| | - Kaiqun Ren
- Department of Pharmaceutical Science, Medical College, Hunan Normal University, Changsha, 410013, Hunan, China. .,Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Changsha, 410013, Hunan, China.
| | - Chang Zou
- Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, Shenzhen People's Hospital, Shenzhen, 518020, China.,Clinical Medical Research Center, the Second Clinical Medical School of Jinan University, Shenzhen People's Hospital, Shenzhen, 518020, China
| | - A Chen
- Department of Pharmaceutical Science, Medical College, Hunan Normal University, Changsha, 410013, Hunan, China.,Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Changsha, 410013, Hunan, China
| | - Chang Xu
- Department of Pharmaceutical Science, Medical College, Hunan Normal University, Changsha, 410013, Hunan, China.,Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Changsha, 410013, Hunan, China
| | - Yebei Qiu
- Department of Pharmaceutical Science, Medical College, Hunan Normal University, Changsha, 410013, Hunan, China.,Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Changsha, 410013, Hunan, China
| | - Meifang Quan
- Department of Pharmaceutical Science, Medical College, Hunan Normal University, Changsha, 410013, Hunan, China.,Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Changsha, 410013, Hunan, China
| | - Jiansong Zhang
- Department of Preclinical Medicine, Medical College, Hunan Normal University, Changsha, 410013, Hunan, China
| | - Jianguo Cao
- Department of Pharmaceutical Science, Medical College, Hunan Normal University, Changsha, 410013, Hunan, China. .,Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Changsha, 410013, Hunan, China.
| | - Xiangding Chen
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China.
| |
Collapse
|
7
|
Abstract
Gastrointestinal malignancies are challenging cancers with considerable economic and societal impacts on health care systems worldwide. While advances in surgical approaches have provided benefits to a proportion of patients, only modest improvements have been attained in the treatment of patients with advanced disease, resulting in limited improvement in survival rates in these patients. Oncolytic adenoviruses are being developed to address gastrointestinal malignancies. Each platform has evolved to maximize tumor-cell killing potency while minimizing toxicities. Tumor-specific bioengineered adenoviruses using chimeric promoters, prodrug convertase enzymes, lethal genes, tumor suppressor genes, and pseudo-typed capsids can provide the innovations for eventual success of oncolytic virotherapy. This article will review the developments in adenoviral platforms in the context of specific gastrointestinal cancers. From the bench to the implementation of clinical trials, this review aims to highlight advances in the field from its early days to the current state of affairs as it pertains to the application of adenoviral oncolytic therapy to gastrointestinal cancers.
Collapse
Affiliation(s)
- Raquel T Yokoda
- Department of Medicine, Division of Hematology Oncology, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ 85205, USA.
| | - Bolni M Nagalo
- Department of Medicine, Division of Hematology Oncology, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ 85205, USA.
| | - Mitesh J Borad
- Department of Medicine, Division of Hematology Oncology, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ 85205, USA.
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA.
- Center for Individualized Medicine, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, USA.
- Mayo Clinic Cancer Center, 5881 E Mayo Blvd, Phoenix, AZ 85054, USA.
| |
Collapse
|
8
|
Ying C, Xiao BD, Qin Y, Wang BR, Liu XY, Wang RW, Fang L, Yan H, Zhou XM, Wang YG. GOLPH2-regulated oncolytic adenovirus, GD55, exerts strong killing effect on human prostate cancer stem-like cells in vitro and in vivo. Acta Pharmacol Sin 2018; 39:405-414. [PMID: 28880012 DOI: 10.1038/aps.2017.91] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 06/05/2017] [Indexed: 12/14/2022]
Abstract
GOLPH2 (also called GP73) is a Golgi glycoprotein, which has been identified as a novel tumor marker upregulated in various cancers, including prostate cancer (PCa). GD55 is a novel GOLPH2-regulated oncolytic adenovirus that exhibits a strong killing effect on hepatoma cells. Here, we investigate the antitumor effect of GD55 on prostate cancer stem cell (CSC)-like cells in vitro and in vivo. Prostate CSC-like sphere cells were acquired and enriched by culturing DU145, LNCap or P3 prostate cancer cells in suspension. The prostate CSC-like sphere cells were capable of self-renewal, differentiation and quiescence, displaying tumorigenic feature and chemo-resistance to 5-FU, doxorubicin and DDP. Treatment with GD55 (1, 5, 10 MOI) dose-dependently suppressed the viability of DU145 sphere cells, which was a more pronounced compared to its cytotoxic action on the parental DU145 cells. In a mouse xenograft prostate CSC-like model, intratumoral injection of GD55 markedly suppressed the growth rate of xenograft tumors and induced higher levels of cell death and necrosis within the tumor tissues. Our results demonstrate that GD55 infection exerts strong anticancer effects on prostate CSC-like cells in vitro and in vivo, and has a potential to be used in the clinical therapy of PCa.
Collapse
|
9
|
Yang M, Yang CS, Guo W, Tang J, Huang Q, Feng S, Jiang A, Xu X, Jiang G, Liu YQ. A novel fiber chimeric conditionally replicative adenovirus-Ad5/F35 for tumor therapy. Cancer Biol Ther 2017; 18:833-840. [PMID: 29144842 DOI: 10.1080/15384047.2017.1395115] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Significant progress has been made in the diagnosis and treatment of cancer; however, significant challenges remain. Conditionally replicating adenoviruses (CRAds), which not only kill cancer cells, but also serve as vectors to express therapeutic genes, are a novel and effective method to treat cancer. However, most adenoviruses are Ad5, which infect cells through the coxsackie and adenovirus receptor (CAR). The transduction efficacy of Ad5 is restricted because of the absent or low expression of CAR on several cancer cells. Ad serotype 35 has a different tropism pattern to Ad5. Ad35 attaches to cells via a non-CAR receptor, CD46, which is expressed widely on most tumor cells. Thus, chimeric adenoviral vectors consisting of the knob and shaft of Ad35 combined with Ad5 have been constructed. The chimeric fiber adenoviral vectors can transduce CAR-positive and CAR-negative cell lines. In this review, we explore the application of the novel fiber chimeric conditionally replicative adenovirus-Ad5/F35 in tumor therapy in terms of safety, mechanism, transduction efficacy, and antitumor effect.
Collapse
Affiliation(s)
- Ming Yang
- a Department of Radiotherapy , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China.,b Department of Oncology , Affiliated Nanyang Second General Hospital , Nanyang , China
| | - Chun Sheng Yang
- c Department of Dermatology , Affiliated Huai'an Hospital of Xuzhou Medical University , the Second People's Hospital of Huai'an, Huai'an , China
| | - WenWen Guo
- a Department of Radiotherapy , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| | - JianQin Tang
- d Department of Dermatology , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| | - Qian Huang
- a Department of Radiotherapy , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| | - ShouXin Feng
- a Department of Radiotherapy , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| | - AiJun Jiang
- a Department of Radiotherapy , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| | - XiFeng Xu
- a Department of Radiotherapy , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| | - Guan Jiang
- d Department of Dermatology , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| | - Yan Qun Liu
- d Department of Dermatology , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| |
Collapse
|
10
|
Hedgehog signaling pathway affects the sensitivity of hepatoma cells to drug therapy through the ABCC1 transporter. J Transl Med 2017; 97:819-832. [PMID: 28414325 DOI: 10.1038/labinvest.2017.34] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 01/18/2017] [Accepted: 02/07/2017] [Indexed: 12/29/2022] Open
Abstract
The poor response to drug therapy often seen in hepatocellular carcinoma requires insight into the molecular interplay responsible for intrinsic or acquired drug resistance. We previously demonstrated that the CD133-/EpCAM- subpopulation of the Huh-7 hepatoma cell line features aberrant activation of the hedgehog signaling (Hh) pathway and chemoresistance. The prevailing hypothesis of the present study is that hedgehog signaling may govern expression of ATP-binding cassette (ABC) transporters, which are responsible for drug resistance in the CD133-/EpCAM- subpopulation. Our aim is to reveal the molecular interplay in the mediation of drug resistance with a newly established Huh-7 subpopulation featuring high Hh signaling activity and drug resistance. In this study, chemoresistance was determined in a newly established Huh-7-DN subpopulation featuring the CD133-/EpCAM- surface marker profile, aberrant expression of Hh pathway, and epithelial-mesenchymal transition (EMT). Expression of ABC transporter proteins (ABCB1, ABCC1, and ABCG2) and Hh transcription factor Gli-1/2 was evaluated with and without Hh signaling antagonists LDE225 or itraconazole. We found that hedgehog signaling activity as determined by transfection with a Gli-Lux reporter cassette and gene expression levels tended to increase from Huh-7 CD133+/EpCAM+ to CD133-/EpCAM-, and the highest levels were found in Huh-7-DN cells. The Huh-7-DN subpopulation exhibited characteristics of EMT as evidenced by increased expression of vimentin and loss of E-cadherin. Sorafenib significantly inhibited the viability of all subpopulations except the Huh-7-DN subpopulation. Compared with other sorafenib-sensitive subpopulations, the Huh-7-DN subpopulation showed enhanced expression of Hh transcription factor Gli-2 and ABCC1 transporter protein. Silencing Gli-2 by lentivirus harboring shRNA against Gli-2 or LDE225 significantly suppressed expression of Gli-2 and ABCC1 genes in Huh-7-DN subpopulation. In conclusion, aberrant hedgehog signaling activation is linked to poor differentiation, epithelial-mesenchymal transition, and chemoresistance in the Huh-7-DN subpopulation. Hedgehog signaling transcription factor Gli-2 appears to be the primary regulator for drug sensitivity of hepatoma through the ABCC1 transporter.
Collapse
|
11
|
Ma X, Yang Y, Li HL, Zheng W, Gao J, Zhang W, Yang G, Shu XO, Xiang YB. Dietary trace element intake and liver cancer risk: Results from two population-based cohorts in China. Int J Cancer 2016; 140:1050-1059. [PMID: 27859272 DOI: 10.1002/ijc.30522] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 10/21/2016] [Accepted: 11/02/2016] [Indexed: 01/07/2023]
Abstract
Dietary factors have been hypothesized to affect the risk of liver cancer via various mechanisms, but the influence has been not well studied and the evidence is conflicting. We investigated associations of dietary trace element intake, assessed through a validated food frequency questionnaire, with risk of liver cancer in two prospective cohort studies of 132,765 women (1997-2013) and men (2002-2013) in Shanghai, China. The associations were first evaluated in cohort studies and further assessed in a case-control study nested within these cohorts adjusting for hepatitis B virus infection. For cohort analyses, Cox proportional hazard models were used to estimate hazard ratios and 95% confidence intervals. For nested case-control analyses, conditional logistic regression was used to calculate odds ratios and 95% confidence intervals. After a median follow-up time of 15.2 years for the Shanghai Women's Health Study and 9.3 years for the Shanghai Men's Health Study, 192 women and 344 men developed liver cancer. Dietary intake of manganese was inversely associated with liver cancer risk (highest vs. lowest quintile, HR = 0.51, 95% CI: 0.35-0.73; ptrend = 0.001). Further adjustment for hepatitis B virus infection in the nested case-control study yielded a similar result (highest vs. lowest quintile, OR = 0.38, 95% CI: 0.21-0.69; ptrend < 0.001). No significant association was found between dietary intake of selenium, iron, zinc, copper and liver cancer risk. The results suggest that higher intake of manganese may be associated with a lower risk of liver cancer in China.
Collapse
Affiliation(s)
- Xiao Ma
- School of Public Health, Fudan University, Shanghai, China.,SKLORG & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yang Yang
- SKLORG & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hong-Lan Li
- SKLORG & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN
| | - Jing Gao
- SKLORG & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wei Zhang
- SKLORG & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Gong Yang
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN
| | - Yong-Bing Xiang
- SKLORG & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
12
|
Wang S, Shu J, Chen L, Chen X, Zhao J, Li S, Mou X, Tong X. Synergistic suppression effect on tumor growth of ovarian cancer by combining cisplatin with a manganese superoxide dismutase-armed oncolytic adenovirus. Onco Targets Ther 2016; 9:6381-6388. [PMID: 27799786 PMCID: PMC5074737 DOI: 10.2147/ott.s113014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Gene therapy on the basis of oncolytic adenovirus is a novel approach for human cancer therapeutics. We aim to investigate whether it will synergistically reinforce their antiovarian cancer activities when the combined use of ZD55-manganese superoxide dismutase (MnSOD) and cisplatin was performed. The experiments in vitro showed that ZD55-MnSOD enhances cisplatin-induced apoptosis and causes remarkable ovarian cancer cell death. Apoptosis induction by treatment with ZD55-MnSOD and/or cisplatin was detected in SKOV-3 by apoptotic cell staining, flow cytometry, and western blot analysis. In addition, the cytotoxicity caused by ZD55-MnSOD to normal cells was examined by the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide assay and western blot analysis. Animal experiment further confirmed that combination of ZD55-MnSOD and cisplatin achieved significant inhibition of SKOV-3 ovarian tumor xenografted growth. In summary, we have demonstrated that ZD55-MnSOD can sensitize human ovarian cancer cells to cisplatin-induced cell death and apoptosis in vitro and in vivo. These findings indicate that the combined treatment with ZD55-MnSOD and cisplatin could represent a rational approach for antiovarian cancer therapy.
Collapse
Affiliation(s)
- Shibing Wang
- Clinical Research Institute, Zhejiang Provincial People's Hospital; Key Laboratory of Cancer Molecular Diagnosis and Individualized Therapy of Zhejiang Province
| | - Jing Shu
- Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital
| | - Li Chen
- Department of Obstetrics and Gynecology, Hangzhou Red Cross Hospital, Hangzhou, People's Republic of China
| | - Xiaopan Chen
- Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital
| | - Jianhong Zhao
- Department of Obstetrics and Gynecology, Hangzhou Red Cross Hospital, Hangzhou, People's Republic of China
| | - Shuangshuang Li
- Clinical Research Institute, Zhejiang Provincial People's Hospital; Key Laboratory of Cancer Molecular Diagnosis and Individualized Therapy of Zhejiang Province
| | - Xiaozhou Mou
- Clinical Research Institute, Zhejiang Provincial People's Hospital; Key Laboratory of Cancer Molecular Diagnosis and Individualized Therapy of Zhejiang Province
| | - Xiangmin Tong
- Clinical Research Institute, Zhejiang Provincial People's Hospital; Key Laboratory of Cancer Molecular Diagnosis and Individualized Therapy of Zhejiang Province
| |
Collapse
|
13
|
Zhang R, Zhang X, Ma B, Xiao B, Huang F, Huang P, Ying C, Liu T, Wang Y. Enhanced antitumor effect of combining TRAIL and MnSOD mediated by CEA-controlled oncolytic adenovirus in lung cancer. Cancer Gene Ther 2016; 23:168-77. [PMID: 27080225 DOI: 10.1038/cgt.2016.11] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 02/14/2016] [Accepted: 02/16/2016] [Indexed: 12/17/2022]
Abstract
Lung cancer, especially adenocarcinoma, is one of the leading causes of death in the world. Carcinoembryonic antigen (CEA), a superb non-small-cell lung cancer marker candidate, showed a beneficial effect in cancer therapy with oncolytic adenovirus in recent studies. Cancer-targeting dual gene-virotherapy delivers two therapeutic genes, linked by a connexon, in the replication-deficient vector instead of one gene so that they can work in common. In this study, we constructed a tumor-specific oncolytic adenovirus, CD55-TRAIL-IETD-MnSOD. The virus has the fusion protein complementary DNAs for tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and for manganese superoxide dismutase (MnSOD) complementary DNA linked through a 4-amino acid caspase-8 cleavage site (IETD), and uses a CEA promoter to control virus E1A express. This is the first work to use a CEA promoter-regulated oncolytic adenovirus carrying two therapeutic genes for cancer research. Its targeting and anticancer capacity was evaluated by in vitro and in vivo experiments. The results indicated that CD55-TRAIL-IETD-MnSOD caused more cell apoptosis than CD55-TRAIL or CD55-MnSOD alone, or their combination in vitro, with low cytotoxicity of normal cells. In the A549 tumor xenograft model in nude mice, data showed that CD55-TRAIL-IETD-MnSOD could effectively suppress tumor growth than single gene groups, with no histological damage in liver, spleen or kidney tissues. Thus, the CEA-regulated dual-gene oncolytic virus CD55-TRAIL-IETD-MnSOD may be a novel potential therapy for lung cancer.
Collapse
Affiliation(s)
- R Zhang
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, People's Republic of China
| | - X Zhang
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, People's Republic of China
| | - B Ma
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, People's Republic of China.,Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - B Xiao
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, People's Republic of China
| | - F Huang
- School of Public health, Zhejiang University, Hangzhou, People's Republic of China
| | - P Huang
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, People's Republic of China
| | - C Ying
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, People's Republic of China
| | - T Liu
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Y Wang
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, People's Republic of China
| |
Collapse
|
14
|
Azzolin VF, Cadoná FC, Machado AK, Berto MD, Barbisan F, Dornelles EB, Glanzner WG, Gonçalves PB, Bica CG, da Cruz IBM. Superoxide-hydrogen peroxide imbalance interferes with colorectal cancer cells viability, proliferation and oxaliplatin response. Toxicol In Vitro 2015; 32:8-15. [PMID: 26674755 DOI: 10.1016/j.tiv.2015.12.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 11/30/2015] [Accepted: 12/02/2015] [Indexed: 10/22/2022]
Abstract
The role of superoxide dismutase manganese dependent enzyme (SOD2) in colorectal cancer is presently insufficiently understood. Some studies suggest that high SOD2 levels found in cancer tissues are associated with cancer progression. However, thus far, the role of colorectal cancer superoxide-hydrogen peroxide imbalance has not yet been studied. Thus, in order to address this gap in extant literature, we performed an in vitro analysis using HT-29 colorectal cell line exposed to paraquat, which generates high superoxide levels, and porphyrin, a SOD2 mimic molecule. The effect of these drugs on colorectal cancer cell response to oxaliplatin was evaluated. At 0.1 μM concentration, both drugs exhibited cytotoxic and antiproliferative effect on colorectal cancer cells. However, this effect was more pronounced in cells exposed to paraquat. Paraquat also augmented the oxaliplatin cytotoxic and antiproliferative effects by increasing the number of apoptosis events, thus causing the cell cycle arrest in the S and M/G2 phases. The treatments were also able to differentially modulate genes related to apoptosis, cell proliferation and antioxidant enzyme system. However, the effects were highly variable and the results obtained were inconclusive. Nonetheless, our findings support the hypothesis that imbalance caused by increased hydrogen peroxide levels could be beneficial to cancer cell biology. Therefore, the use of therapeutic strategies to decrease hydrogen peroxide levels mainly during oxaliplatin chemotherapy could be clinically important to the outcomes of colorectal cancer treatment.
Collapse
Affiliation(s)
- Verônica Farina Azzolin
- Programa de Pós-Graduação em Farmacologia, Centro de Ciências da Saúde, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Francine Carla Cadoná
- Programa de Pós-Graduação em Bioquímica Toxicológica, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Alencar Kolinski Machado
- Programa de Pós-Graduação em Farmacologia, Centro de Ciências da Saúde, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | | | - Fernanda Barbisan
- Programa de Pós-Graduação em Farmacologia, Centro de Ciências da Saúde, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Eduardo Bortoluzzi Dornelles
- Programa de Pós-Graduação em Bioquímica Toxicológica, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Werner Giehl Glanzner
- BIOREP Lab, Centro de Ciências Agrárias, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Paulo Bayard Gonçalves
- BIOREP Lab, Centro de Ciências Agrárias, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | | | - Ivana Beatrice Mânica da Cruz
- Programa de Pós-Graduação em Farmacologia, Centro de Ciências da Saúde, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil; Programa de Pós-Graduação em Bioquímica Toxicológica, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil.
| |
Collapse
|
15
|
Yaghchi CA, Zhang Z, Alusi G, Lemoine NR, Wang Y. Vaccinia virus, a promising new therapeutic agent for pancreatic cancer. Immunotherapy 2015; 7:1249-58. [PMID: 26595180 PMCID: PMC4976866 DOI: 10.2217/imt.15.90] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The poor prognosis of pancreatic cancer patients signifies a need for radically new therapeutic strategies. Tumor-targeted oncolytic viruses have emerged as attractive therapeutic candidates for cancer treatment due to their inherent ability to specifically target and lyse tumor cells as well as induce antitumor effects by multiple action mechanisms. Vaccinia virus has several inherent features that make it particularly suitable for use as an oncolytic agent. In this review, we will discuss the potential of vaccinia virus in the management of pancreatic cancer in light of our increased understanding of cellular and immunological mechanisms involved in the disease process as well as our extending knowledge in the biology of vaccinia virus.
Collapse
Affiliation(s)
- Chadwan Al Yaghchi
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, UK
| | - Zhongxian Zhang
- National Centre for International Research in Cell & Gene Therapy, Sino-British Research Centre for Molecular Oncology, Zhengzhou University, China
| | - Ghassan Alusi
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, UK
| | - Nicholas R Lemoine
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, UK
- National Centre for International Research in Cell & Gene Therapy, Sino-British Research Centre for Molecular Oncology, Zhengzhou University, China
| | - Yaohe Wang
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, UK
- National Centre for International Research in Cell & Gene Therapy, Sino-British Research Centre for Molecular Oncology, Zhengzhou University, China
| |
Collapse
|
16
|
Huang F, Ma B, Wang Y, Xiao R, Kong Y, Zhou X, Xia D. Targeting gene-virus-mediated manganese superoxide dismutase effectively suppresses tumor growth in hepatocellular carcinoma in vitro and in vivo. Cancer Biother Radiopharm 2015; 29:403-11. [PMID: 25414976 DOI: 10.1089/cbr.2014.1642] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Although the treatment methods for hepatocellular carcinoma (HCC) have made a great progress on patient survival rate and life quality, the HCC recurrence still is very high. To explore the novel effective anticancer strategies for HCC, the Cancer Targeting Gene-Viro-Therapy (CTGVT) strategy was applied through oncolytic virus-delivery antitumor gene. In this article, the dual-regulated oncolytic adenovirus Ad-AFP-E1A-E1B(Δ55kDa)-Mn-SOD (briefly named AD55-Mn-SOD) was constructed using a liver cancer-specific α-fetoprotein (AFP) promoter to control replication-essential E1A gene and deliver the novel tumor suppression gene Manganese superoxide dismutase (Mn-SOD). The results indicated that the constructed AD55-Mn-SOD exerted tumor-specific features, and induced dramatic cytotoxicity in HCC cells in vitro and suppress the HCC xenografted growth in nude mice. Moreover, the anticancer mechanism of AD55-Mn-SOD is due to the activation of caspase apoptotic pathway. These data suggested that AD55-Mn-SOD could become a potential anticancer agent for liver cancer.
Collapse
Affiliation(s)
- Fang Huang
- 1 School of Public Health, Zhejiang University , Hangzhou, China
| | | | | | | | | | | | | |
Collapse
|
17
|
Wang Y, Liu T, Huang P, Zhao H, Zhang R, Ma B, Chen K, Huang F, Zhou X, Cui C, Liu X. A novel Golgi protein (GOLPH2)-regulated oncolytic adenovirus exhibits potent antitumor efficacy in hepatocellular carcinoma. Oncotarget 2015; 6:13564-78. [PMID: 25980438 PMCID: PMC4537034 DOI: 10.18632/oncotarget.3769] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 04/10/2015] [Indexed: 12/16/2022] Open
Abstract
Golgi apparatus is the organelle mainly functioning as protein processing and secretion. GOLPH2 is a resident Golgi glycoprotein, usually called GP73. Recent data displayed that GOLPH2 is a superb hepatocellular carcinoma (HCC) marker candidate, and even its specificity is better than liver cancer marker AFP. Oncolytic adenoviruses are broadly used for targeting cancer therapy due to their selective tumor-killing effect. However, it was reported that traditionally oncolytic adenovirus lack the HCC specificity. In this study, a novel dual-regulated oncolytic adenovirus GD55 targeting HCC was first constructed based on our cancer targeted gene-viral therapeutic strategy. To verify the targeting and effectiveness of GOLPH2-regulated oncolytic adenovirus GD55 in HCC, the anticancer capacity was investigated in HCC cell lines and animal model. The results proved that the novel GOLPH2-regulated GD55 conferred higher adenovirus replication and infectivity for liver cancer cells than oncolytic adenovirus ZD55. The GOLPH2-regulated GD55 exerted a significant grow-suppressing effect on HCC cells in vitro but little damage to normal liver cells. In animal experiment, antitumor effect of GD55 was more effective in HCC xenograft of nude mice than that of ZD55. Thus GOLPH2-regulated GD55 may be a promising oncolytic virus agent for future liver cancer treatment.
Collapse
Affiliation(s)
- Yigang Wang
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, PR China
| | - Tao Liu
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, PR China
| | - Panpan Huang
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, PR China
| | - Hongfang Zhao
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, PR China
| | - Rong Zhang
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, PR China
| | - Buyun Ma
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, PR China
| | - Kan Chen
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, PR China
| | - Fang Huang
- School of Public Health, Zhejiang University, Hangzhou 310058, PR China
| | - Xiumei Zhou
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, PR China
| | - Caixia Cui
- Otorhinolaryngology Head and Neck Surgery, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, PR China
| | - Xinyuan Liu
- Xinyuan Institute of Medicine and Biotechnology, School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, PR China.,Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, PR China
| |
Collapse
|
18
|
Han X, Wang S, Zhou W, Li Y, Lei W, Lv W. Synergistic combination of histone deacetylase inhibitor suberoylanilide hydroxamic acid and oncolytic adenovirus ZD55-TRAIL as a therapy against cervical cancer. Mol Med Rep 2015; 12:435-41. [PMID: 25684632 DOI: 10.3892/mmr.2015.3355] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 01/21/2015] [Indexed: 11/06/2022] Open
Abstract
Oncolytic adenoviruses (OA) have been investigated as virotherapeutic agents for the treatment of cervical cancer and thus far results are promising. However, the cytotoxicity of the viruses requires improvement. The present study demonstrated that this can be achieved by combining ZD55-TRAIL, an OA containing the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) gene, with the histone deacetylase inhibitor suberoylanilide hydroxamic acid (SAHA). It was demonstrated that these agents act synergistically to kill HeLa cells by inducing G2 growth arrest and apoptosis. Notably, in a mouse xenograft model, ZD55-TRAIL/SAHA combination inhibited tumor growth. At the molecular level, it was found that upregulation of IκBα and the p50 and p65 subunits of nuclear factor-κB induced by ZD55-TRAIL, can be abrogated by SAHA treatment. These data strongly suggested that ZD55-TRAIL/SAHA co-treatment may serve as an effective therapeutic strategy against cervical cancer.
Collapse
Affiliation(s)
- Xiujun Han
- Institute of Oncology, Women's Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Shibing Wang
- Xinyuan Institute of Medicine and Biotechnology, School of Life Science, Zhejiang Sci‑Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | - Wenjing Zhou
- Institute of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Ying Li
- Institute of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Wen Lei
- Institute of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Weiguo Lv
- Institute of Oncology, Women's Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
19
|
Hosoya N, Sakumoto M, Tomita Y, Kondo T. Approach to spot overlapping problem in 2D-PAGE revealed clinical and functional significance of RKIP and MnSOD in renal cell carcinoma. EUPA OPEN PROTEOMICS 2014. [DOI: 10.1016/j.euprot.2014.06.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
20
|
Pan Q, Huang Y, Chen L, Gu J, Zhou X. SMAC-armed vaccinia virus induces both apoptosis and necroptosis and synergizes the efficiency of vinblastine in HCC. Hum Cell 2014; 27:162-71. [PMID: 24771354 DOI: 10.1007/s13577-014-0093-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 03/24/2014] [Indexed: 12/14/2022]
Abstract
Hepatocellular carcinoma (HCC) has particularly high incidence rate in Asia and its resistance to the chemotherapeutic drugs and cell death make it intractable. Vaccinia virus (VV) is a potential vehicle and has been widely used in cancer therapy. SMAC/DIABLO is a critical factor in activating caspases and eliminating inhibition of IAPs when the programmed cell death is promoted. In this study, we constructed a tumor-targeted vaccinia virus carrying SMAC/DIABLO gene that was knocked in the region of viral thymidine kinase gene (VV-SMAC). Our results showed that VV-SMAC efficiently infected and destroyed HCC cells via triggering both caspase-dependent apoptosis and necroptosis with depletion of IAPs. Furthermore, ripoptosome, a prerequisite complex of necroptosis, was assembled and induced by VV-SMAC. In addition, the combination of VV-SMAC and vinblastine represented a synergistic effect on HCC cells. In summary, our data suggest that VV-SMAC is a potential candidate and combination of VV-SMAC and vinblastine may provide a new avenue in treatment of HCC.
Collapse
Affiliation(s)
- Qiang Pan
- Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Room 217, Building No. 6, Xiasha, Hangzhou, 310018, China
| | | | | | | | | |
Collapse
|
21
|
Abstract
SIGNIFICANCE Cancer is the second leading cause of death in the United States. Considering the quality of life and treatment cost, the best way to fight against cancer is to prevent or suppress cancer development. Cancer is preventable as indicated by human papilloma virus (HPV) vaccination and tamoxifen/raloxifen treatment in breast cancer prevention. The activities of superoxide dismutases (SODs) are often lowered during early cancer development, making it a rational candidate for cancer prevention. RECENT ADVANCES SOD liposome and mimetics have been shown to be effective in cancer prevention animal models. They've also passed safety tests during early phase clinical trials. Dietary supplement-based SOD cancer prevention provides another opportunity for antioxidant-based cancer prevention. New mechanistic studies have revealed that SOD inhibits not only oncogenic activity, but also subsequent metabolic shifts during early tumorigenesis. CRITICAL ISSUES Lack of sufficient animal model studies targeting specific cancers; and lack of clinical trials and support from pharmaceutical industries also hamper efforts in further advancing SOD-based cancer prevention. FUTURE DIRECTIONS To educate and obtain support from our society that cancer is preventable. To combine SOD-based therapeutics with other cancer preventive agents to obtain synergistic effects. To formulate a dietary supplementation-based antioxidant approach for cancer prevention. Lastly, targeting specific populations who are prone to carcinogens, which can trigger oxidative stress as the mechanism of carcinogenesis.
Collapse
Affiliation(s)
- Delira Robbins
- 1 Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital , Memphis, Tennessee
| | | |
Collapse
|
22
|
Li G, Li X, Wu H, Yang X, Zhang Y, Chen L, Wu X, Cui L, Wu L, Luo J, Liu XY. CD123 targeting oncolytic adenoviruses suppress acute myeloid leukemia cell proliferation in vitro and in vivo. Blood Cancer J 2014; 4:e194. [PMID: 24658372 PMCID: PMC3972701 DOI: 10.1038/bcj.2014.15] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 02/14/2014] [Indexed: 12/21/2022] Open
Abstract
We report here a novel strategy to redirect oncolytic adenoviruses to CD123 by carry a soluble coxsackie-adenovirus receptor (sCAR)-IL3 expression cassette in the viral genome to form Ad.IL3, which sustainably infected acute myeloid leukemia (AML) cells through CD123. Ad.IL3 was further engineered to harbor gene encoding manganese superoxide dismutase (MnSOD) or mannose-binding plant lectin Pinellia pedatisecta agglutinin (PPA), forming Ad.IL3-MnSOD and Ad.IL3-PPA. As compared with Ad.IL3 or Ad.sp-E1A control, Ad.IL3-MnSOD and Ad.IL3-PPA significantly suppressed in vitro proliferation of HL60 and KG-1 cells. Elevated apoptosis was detected in HL60 and KG-1 cells treated with either Ad.IL3-MnSOD or Ad.IL3-PPA. The caspase-9–caspase-7 pathway was determined to be activated by Ad.IL3-MnSOD as well as by Ad.IL3-PPA in HL60 cells. In an HL60/Luc xenograft nonobese diabetic/severe-combined immunodeficiency mice model, Ad.IL3-MnSOD and Ad.IL3-PPA suppressed cancer cell growth as compared with Ad.IL3. A significant difference of cancer cell burden was detected between Ad.IL3 and Ad.IL3-PPA groups at day 9 after treatment. Furthermore, Ad.IL3-MnSOD significantly prolonged mouse survival as compared with Ad.sp-E1A. These findings demonstrated that Ad.IL3-gene could serve as a novel agent for AML therapy. Harboring sCAR-ligand expression cassette in the viral genome may provide a universal method to redirect oncolytic adenoviruses to various membrane receptors on cancer cells resisting serotype 5 adenovirus infection.
Collapse
Affiliation(s)
- G Li
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - X Li
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - H Wu
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - X Yang
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - Y Zhang
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - L Chen
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - X Wu
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - L Cui
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - L Wu
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - J Luo
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - X Y Liu
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| |
Collapse
|
23
|
Policastro LL, Ibañez IL, Notcovich C, Duran HA, Podhajcer OL. The tumor microenvironment: characterization, redox considerations, and novel approaches for reactive oxygen species-targeted gene therapy. Antioxid Redox Signal 2013; 19:854-95. [PMID: 22794113 DOI: 10.1089/ars.2011.4367] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The tumor microenvironment is a complex system that involves the interaction between malignant and neighbor stromal cells embedded in a mesh of extracellular matrix (ECM) components. Stromal cells (fibroblasts, endothelial, and inflammatory cells) are co-opted at different stages to help malignant cells invade the surrounding ECM and disseminate. Malignant cells have developed adaptive mechanisms to survive under the extreme conditions of the tumor microenvironment such as restricted oxygen supply (hypoxia), nutrient deprivation, and a prooxidant state among others. These conditions could be eventually used to target drugs that will be activated specifically in this microenvironment. Preclinical studies have shown that modulating cellular/tissue redox state by different gene therapy (GT) approaches was able to control tumor growth. In this review, we describe the most relevant features of the tumor microenvironment, addressing reactive oxygen species-generating sources that promote a prooxidative microenvironment inside the tumor mass. We describe different GT approaches that promote either a decreased or exacerbated prooxidative microenvironment, and those that make use of the differential levels of ROS between cancer and normal cells to achieve tumor growth inhibition.
Collapse
Affiliation(s)
- Lucia Laura Policastro
- Department of Micro and Nanotechnology, National Atomic Energy Commission, Buenos Aires 1650, Argentina.
| | | | | | | | | |
Collapse
|
24
|
Meng X, Wu J, Pan C, Wang H, Ying X, Zhou Y, Yu H, Zuo Y, Pan Z, Liu RY, Huang W. Genetic and epigenetic down-regulation of microRNA-212 promotes colorectal tumor metastasis via dysregulation of MnSOD. Gastroenterology 2013; 145:426-36.e1-6. [PMID: 23583431 DOI: 10.1053/j.gastro.2013.04.004] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2012] [Revised: 03/22/2013] [Accepted: 04/03/2013] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS Altered functions of microRNAs (miRNAs) have been associated with colorectal cancer (CRC). miR-212 is transcribed from a stable intron of a non-protein coding gene, and is reportedly down-regulated in different tumor types. We investigated the role of miR-212 in colorectal carcinogenesis and progression. METHODS We analyzed the expression of miR-212 by real-time polymerase chain reaction (PCR) analysis of colorectal cell lines and 180 paired tumor samples and surrounding healthy tissue. We overexpressed and knocked down miR-212 in CRC cell lines and assessed the in vitro effects. We also studied the effects of miR-212 overexpression on metastasis of tumors grown from HCT116 cells in nude mice. RESULTS Overexpression of miR-212 inhibited CRC cell migration and invasion in vitro and formation of intrahepatic and pulmonary metastasis in vivo. We identified manganese superoxide dismutase (MnSOD) messenger RNA as a direct target of miR-212, and observed an inverse correlation between the level of miR-212 and MnSOD protein in colorectal tumor samples. MnSOD was required for down-regulation of epithelial markers and up-regulation of mesenchymal markers in CRC cells, indicating that it promoted the epithelial-mesenchymal transition. Overexpression of miR-212 reduced the levels of MnSOD to block the epithelial-mesenchymal transition process. Loss of heterozygosity and promoter hypermethylation each contributed to the down-regulation of miR-212. Reduced levels of miR-212 were associated with a more aggressive tumor phenotype and short disease-free survival times of patients (P = .0045; overall survival, P = .0015). CONCLUSIONS miR-212 is down-regulated in human CRC tissues via genetic and epigenetic mechanisms. miR-212 might prevent tumor progression by targeting MnSOD messenger RNA; reduction of miR-212 could be a prognostic marker for patients with CRC. miR-212 and MnSOD might also be therapeutic targets for cancer.
Collapse
Affiliation(s)
- Xiangqi Meng
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Elias-Miró M, Jiménez-Castro MB, Rodés J, Peralta C. Current knowledge on oxidative stress in hepatic ischemia/reperfusion. Free Radic Res 2013; 47:555-568. [PMID: 23738581 DOI: 10.3109/10715762.2013.811721] [Citation(s) in RCA: 150] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ischemia/reperfusion (I/R) injury associated with hepatic resections and liver transplantation remains a serious complication in clinical practice, despite several attempts to solve the problem. The redox balance, which is pivotal for normal function and integrity of tissues, is dysregulated during I/R, leading to an accumulation of reactive oxygen species (ROS). Formation of ROS and oxidant stress are the disease mechanisms most commonly invoked in hepatic I/R injury. The present review examines published results regarding possible sources of ROS and their effects in the context of I/R injury. We also review the effect of oxidative stress on marginal livers, which are more vulnerable to I/R-induced oxidative stress. Strategies to improve the viability of marginal livers could reduce the risk of dysfunction after surgery and increase the number of organs suitable for transplantation. The review also considers the therapeutic strategies developed in recent years to reduce the oxidative stress induced by hepatic I/R, and we seek to explain why some of them have not been applied clinically. New antioxidant strategies that have yielded promising results for hepatic I/R injury are discussed.
Collapse
Affiliation(s)
- M Elias-Miró
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona (IDIBAPS), Spain
| | | | | | | |
Collapse
|
26
|
Zhou X, Xie G, Wang S, Wang Y, Zhang K, Zheng S, Chu L, Xiao L, Yu Y, Zhang Y, Liu X. Potent and specific antitumor effect for colorectal cancer by CEA and Rb double regulated oncolytic adenovirus harboring ST13 gene. PLoS One 2012; 7:e47566. [PMID: 23077639 PMCID: PMC3471845 DOI: 10.1371/journal.pone.0047566] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 09/18/2012] [Indexed: 01/19/2023] Open
Abstract
Cancer Targeting Gene-Viro-Therapy (CTGVT) is constructed by inserting an antitumor gene into an oncolytic virus (OV). It is actually an OV-gene therapy, which has much better antitumor effect than either gene therapy alone or virotherapy alone in our previously published papers. This study is a modification of CTGVT by inserting a colorectal cancer (CRC) specific suppressor gene, ST13, into a CRC specific oncolytic virus, the Ad·CEA·E1A(Δ24), to construct the Ad·(ST13)·CEA·E1A(Δ24) for increasing the targeting tropism to colorectal cancer and it was briefly named as CTGVT-CRC. Although many studies on CEA promoter and ST13 gene were reported but no construct has been performed to combine both of them as a new strategy for colorectal cancer (CRC) specific therapy. In addition to the CRC specificity, the antitumor effect of Ad·(ST13)·CEA·E1A(Δ24) was also excellent and got nearly complete inhibition (not eradication) of CRC xenograft since ST13 was an effective antitumor gene with less toxicity, and a Chinese patent (No. 201110319434.4) was available for this study. Ad·(ST13)·CEA·E1A(Δ24) caused cell apoptosis through P38 MAPK (i.e. P38) which upregulated CHOP and ATF2 expression. The mitochondrial medicated apoptosis pathway was activated by the increase of caspase 9 and caspase 3 expression.
Collapse
Affiliation(s)
- Xiumei Zhou
- Xinyuan Institute of Medicine and Biotechnology, College of Life Science, Zhejiang Sci-Tech University, Hangzhou, China
| | - Guoliang Xie
- Department of Laboratory Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Shibing Wang
- Xinyuan Institute of Medicine and Biotechnology, College of Life Science, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yigang Wang
- Xinyuan Institute of Medicine and Biotechnology, College of Life Science, Zhejiang Sci-Tech University, Hangzhou, China
| | - Kangjian Zhang
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shu Zheng
- Cancer Institute, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Liang Chu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Lianli Xiao
- Xinyuan Institute of Medicine and Biotechnology, College of Life Science, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yuemei Yu
- Xinyuan Institute of Medicine and Biotechnology, College of Life Science, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yue Zhang
- Xinyuan Institute of Medicine and Biotechnology, College of Life Science, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xinyuan Liu
- Xinyuan Institute of Medicine and Biotechnology, College of Life Science, Zhejiang Sci-Tech University, Hangzhou, China
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
27
|
Wang SB, Tan Y, Lei W, Wang YG, Zhou XM, Jia XY, Zhang KJ, Chu L, Liu XY, Qian WB. Complete eradication of xenograft hepatoma by oncolytic adenovirus ZD55 harboring TRAIL-IETD-Smac gene with broad antitumor effect. Hum Gene Ther 2012; 23:992-1002. [PMID: 22530834 DOI: 10.1089/hum.2011.159] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Cancer-targeting dual-gene virotherapy (CTGVT-DG) is an important modification of CTGVT, in which two suitable genes are used to obtain an excellent antitumor effect. A key problem is to join the two genes to form one fused gene, and then to clone it into the oncolytic viral vector so that only one investigational new drug application, instead of two, is required for clinical use. Many linkers (e.g., internal ribosome entry site) are used to join two genes together, but they are not all equally efficacious. Here, we describe finding the best linker, that is, sequence encoding the four amino acids IETD, to join the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) gene and the second mitochondria-derived activator of caspase (Smac) gene to form TRAIL-IETD-Smac and inserting it into oncolytic viral vector ZD55 to construct ZD55-TRAIL-IETD-Smac, which matched ZD55-TRAIL plus ZD55-Smac in completely eliminating xenograft hepatoma. ZD55-TRAIL-IETD-Smac works by quantitative cleavage at IETD↓by inducing caspase-8; activation or inhibition of caspase-8 could up- or downregulate cleavage, respectively. The cleaved product, TRAIL-IETD, does not affect the function of TRAIL. Numerous experiments have shown that the combined use of ZD55-TRAIL plus ZD55-X could completely eradicate many xenograft tumors, and therefore the IETD is potentially a useful linker to construct many antitumor drugs, for example, ZD55-TRAIL-IETD-X, where X has a compensative or synergetic effect on TRAIL. We found that the antitumor effect of ZD55-IL-24-IETD-TRAIL also has an equivalent antitumor effect compared with the combined use of ZD55-IL-24 plus ZD55-TRAIL, because ZD55-IL-24 could also induce caspase-8. This means that IETD, as a two-gene linker, may have broad use.
Collapse
Affiliation(s)
- Shi-Bing Wang
- Xinyuan Institute of Medicine and Biotechnology, School of Life Science, Zhejiang Sci-Tech University, Hangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Cai Y, Liu X, Huang W, Zhang K, Liu XY. Synergistic antitumor effect of TRAIL and IL-24 with complete eradication of hepatoma in the CTGVT-DG strategy. Acta Biochim Biophys Sin (Shanghai) 2012; 44:535-43. [PMID: 22635106 DOI: 10.1093/abbs/gms031] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The ZD55-tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and ZD55-interleukin (IL)-24 were constructed by inserting TRAIL or IL-24 gene separately into the oncolytic adenovirus named ZD55 (with adenovirus E1B-55kD deletion). The resulting ZD55-TRAIL and ZD55-IL-24 were used in combination to treat xenograft tumors in nude mice model. The results showed that it can not only completely eliminate BEL7404 hepatoma xenograft but also have excellent antitumor effect against gaster, lung, prostate, and breast carcinomas. It was also found that ZD55-TRAIL could not only suppress the tumor growth promoting effect by ZD55-IL-24 at lower dosage, but also substantially reduce the cancer cell viability in their combined use. This is because ZD55-IL-24 and ZD55-TRAIL could mutually enhance each other's antitumor effect greatly. All these findings conspicuously showed the synergistic antitumor effect of TRAIL and IL-24, which is also the reason for the antitumor effect by the combined use of TRAIL and IL-24 in vitro and also in vivo.
Collapse
Affiliation(s)
- Ying Cai
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | | | | |
Collapse
|
29
|
Xu B, Zheng WY, Jin DY, Wang DS, Liu XY, Qin XY. Treatment of pancreatic cancer using an oncolytic virus harboring the lipocalin-2 gene. Cancer 2012; 118:5217-26. [PMID: 22517373 DOI: 10.1002/cncr.27535] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2011] [Revised: 02/05/2012] [Accepted: 02/21/2012] [Indexed: 12/21/2022]
Abstract
BACKGROUND The 5-year survival rate for patients with pancreatic cancer is <5%, and it is always resistant to the current chemoradiotherapy. Therefore, new, effective agents for the treatment of pancreatic cancer are urgently needed. The promising strategy of cancer-targeting gene virotherapy (CTGVT) has demonstrated great anticancer potential. The objective of the current study was to determine whether 1 CTGVT approach, oncolytic virus (OV)-harboring lipocalin-2, is capable of treating pancreatic cancer. METHODS Tissue microarrays were constructed to detect the expression of lipocalin-2 in 60 specimens of pancreatic adenocarcinoma. The clinical significance of lipocalin-2 was investigated in an analysis of correlations between lipocalin-2 expression and matched clinical characteristics. A lipocalin-2-expressing OV, ZD55-lipocalin-2, was constructed by deleting the adenoviral protein E1B55kD. The antitumor efficacy and mechanisms of the OV were investigated in pancreatic cancer cells with v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations in vitro and in vivo. RESULTS Lipocalin-2 expression was correlated with a good prognosis in patients with pancreatic adenocarcinoma. ZD55-lipocalin-2 dramatically inhibited the growth of pancreatic cancer in vitro and in vivo by inducing cytolysis and caspase-dependent apoptosis. CONCLUSIONS Higher lipocalin-2 expression predicted a better prognosis in patients with pancreatic cancer. The results indicated that ZD55-lipocalin-2, which specifically expressed higher levels of lipocalin-2 in tumor cells, may serve as a potent anticancer drug for pancreatic cancer therapy, especially for patients who have pancreatic adenocarcinoma with KRAS mutations.
Collapse
Affiliation(s)
- Bin Xu
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | | | | | | | | | | |
Collapse
|
30
|
Bianchi L, Bruzzese F, Leone A, Gagliardi A, Puglia M, Di Gennaro E, Rocco M, Gimigliano A, Pucci B, Armini A, Bini L, Budillon A. Proteomic analysis identifies differentially expressed proteins after HDAC vorinostat and EGFR inhibitor gefitinib treatments in Hep-2 cancer cells. Proteomics 2012; 11:3725-42. [PMID: 21761561 DOI: 10.1002/pmic.201100092] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Several solid tumors are characterized by poor prognosis and few effective treatment options, other than palliative chemotherapy in the recurrent/metastatic setting. Epidermal growth factor receptor (EGFR) has been considered an important anticancer target because it is involved in the development and progression of several solid tumors; however, only a subset of patients show a clinically meaningful response to EGFR inhibition, particularly to EGFR tyrosine kinase inhibitors such as gefitinib. We have recently demonstrated synergistic antitumor effect of the histone deacetylase inhibitor vorinostat combined with gefitinib. To further characterize the interaction between these two agents, cellular extracts from Hep-2 cancer cells that were untreated or treated for 24 h with either vorinostat or gefitinib alone or with a vorinostat/gefitinib combination were analyzed using 2-D DIGE. Software analysis using DeCyder was performed, and numerous differentially expressed protein spots were visualized between the four examined settings. Using MALDI-TOF MS and ESI-Ion trap MS/MS, several differentially expressed proteins were identified; some of these were validated by Western blotting. Finally, a pathway analysis of experimental data performed using MetaCore highlighted a relevant relationship between the identified proteins and additional potential effectors. In conclusion, we performed a comprehensive analysis of proteins regulated by vorinostat and gefitinib, alone and in combination, providing a useful insight into their mechanisms of action as well as their synergistic interaction.
Collapse
Affiliation(s)
- Laura Bianchi
- Functional Proteomics Laboratory, Department of Molecular Biology, University of Siena, Siena, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Holley AK, Dhar SK, Xu Y, St. Clair DK. Manganese superoxide dismutase: beyond life and death. Amino Acids 2012; 42:139-58. [PMID: 20454814 PMCID: PMC2975048 DOI: 10.1007/s00726-010-0600-9] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Accepted: 04/16/2010] [Indexed: 02/07/2023]
Abstract
Manganese superoxide dismutase (MnSOD) is a nuclear-encoded antioxidant enzyme that localizes to the mitochondria. Expression of MnSOD is essential for the survival of aerobic life. Transgenic mice expressing a luciferase reporter gene under the control of the human MnSOD promoter demonstrate that the level of MnSOD is reduced prior to the formation of cancer. Overexpression of MnSOD in transgenic mice reduces the incidences and multiplicity of papillomas in a DMBA/TPA skin carcinogenesis model. However, MnSOD deficiency does not lead to enhanced tumorigenicity of skin tissue similarly treated because MnSOD can modulate both the p53-mediated apoptosis and AP-1-mediated cell proliferation pathways. Apoptosis is associated with an increase in mitochondrial levels of p53 suggesting a link between MnSOD deficiency and mitochondrial-mediated apoptosis. Activation of p53 is preventable by application of a SOD mimetic (MnTE-2-PyP(5+)). Thus, p53 translocation to mitochondria and subsequent inactivation of MnSOD explain the observed mitochondrial dysfunction that leads to transcription-dependent mechanisms of p53-induced apoptosis. Administration of MnTE-2-PyP(5+) following apoptosis but prior to proliferation leads to suppression of protein carbonyls and reduces the activity of AP-1 and the level of the proliferating cellular nuclear antigen, without reducing the activity of p53 or DNA fragmentation following TPA treatment. Remarkably, the incidence and multiplicity of skin tumors are drastically reduced in mice that receive MnTE-2-PyP(5+) prior to cell proliferation. The results demonstrate the role of MnSOD beyond its essential role for survival and suggest a novel strategy for an antioxidant approach to cancer intervention.
Collapse
Affiliation(s)
| | | | - Yong Xu
- University of Kentucky, Lexington, USA
| | | |
Collapse
|
32
|
Xu HN, Huang WD, Cai Y, Ding M, Gu JF, Wei N, Sun LY, Cao X, Li HG, Zhang KJ, Liu XR, Liu XY. HCCS1-armed, quadruple-regulated oncolytic adenovirus specific for liver cancer as a cancer targeting gene-viro-therapy strategy. Mol Cancer 2011; 10:133. [PMID: 22040050 PMCID: PMC3222618 DOI: 10.1186/1476-4598-10-133] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2010] [Accepted: 11/01/2011] [Indexed: 12/03/2022] Open
Abstract
Background In previously published studies, oncolytic adenovirus-mediated gene therapy has produced good results in targeting cancer cells. However, safety and efficacy, the two most important aspects in cancer therapy, remain serious challenges. The specific expression or deletion of replication related genes in an adenovirus has been frequently utilized to regulate the cancer cell specificity of a virus. Accordingly, in this study, we deleted 24 bp in E1A (bp924-bp947) and the entirety of E1B, including those genes encoding E1B 55kDa and E1B19kDa. We used the survivin promoter (SP) to control E1A in order to construct a new adenovirus vector named Ad.SP.E1A(Δ24).ΔE1B (briefly Ad.SPDD). HCCS1 (hepatocellular carcinoma suppressor 1) is a novel tumor suppressor gene that is able to specifically induce apoptosis in cancer cells. The expression cassette AFP-HCCS1-WPRE-SV40 was inserted into Ad.SPDD to form Ad.SPDD-HCCS1, enabling us to improve the safety and efficacy of oncolytic-mediated gene therapy for liver cancer. Results Ad.SPDD showed a decreased viral yield and less toxicity in normal cells but enhanced toxicity in liver cancer cells, compared with the cancer-specific adenovirus ZD55 (E1B55K deletion). Ad.SPDD-HCCS1 exhibited a potent anti-liver-cancer ability and decreased toxicity in vitro. Ad.SPDD-HCCS1 also showed a measurable capacity to inhibit Huh-7 xenograft tumor growth on nude mice. The underlying mechanism of Ad.SPDD-HCCS1-induced liver cancer cell death was found to be via the mitochondrial apoptosis pathway. Conclusions These results demonstrate that Ad.SPDD-HCCS1 was able to elicit reduced toxicity and enhanced efficacy both in vitro and in vivo compared to a previously constructed oncolytic adenovirus. Ad.SPDD-HCCS1 could be a promising candidate for liver cancer therapy.
Collapse
Affiliation(s)
- Hai-Neng Xu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Liu X, Cao X, Wei R, Cai Y, Li H, Gui J, Zhong D, Liu XY, Huang K. Gene-viro-therapy targeting liver cancer by a dual-regulated oncolytic adenoviral vector harboring IL-24 and TRAIL. Cancer Gene Ther 2011; 19:49-57. [PMID: 21979578 DOI: 10.1038/cgt.2011.67] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cancer-targeting gene-viro-therapy is a promising cancer therapeutic strategy that strengthens the antitumor effect of oncolytic viruses by expressing an inserted foreign antitumor gene. To achieve liver cancer targeting and to improve the safety of the ZD55 vector (a widely-used E1B55KD gene-deleted oncolytic adenoviral vector (OV), we previously constructed), we designed a novel OV named Ad·AFP·D55 that selectively replicates in hepatocellular carcinoma (HCC) cells by replacing the E1A promoter with the liver-cancer specific α-Fetoprotein (AFP) promoter based on the ZD55 vector. We found that the oncolytic adenoviruses Ad·AFP·D55-IL-24 and Ad·AFP·D55-TRAIL express tumor-suppressor gene interleukin-24 (IL-24) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), respectively, significantly suppressed the HCC cell growth in vitro by inducing apoptosis by the caspase-8 and mitochondria-dependent caspase-9 signaling pathways. Furthermore, the combined treatment of Ad·AFP·D55-IL-24 and Ad·AFP·D55-TRAIL showed strong antitumor effects in vivo by significantly inhibiting the tumor growth in HCC HuH-7 cell xenograft mice, and markedly increasing animal survival rate. Therefore, this novel HCC cell-targeting OV carrying tumor-suppressor genes may provide a promising approach for liver cancer gene therapy.
Collapse
Affiliation(s)
- X Liu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Wei RC, Cao X, Gui JH, Zhou XM, Zhong D, Yan QL, Huang WD, Qian QJ, Zhao FL, Liu XY. Augmenting the antitumor effect of TRAIL by SOCS3 with double-regulated replicating oncolytic adenovirus in hepatocellular carcinoma. Hum Gene Ther 2011; 22:1109-19. [PMID: 21361790 DOI: 10.1089/hum.2010.219] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aberrant JAK/STAT3 pathway has been reported to be related to hepatocellular carcinoma (HCC) in many cell lines. In this study, a double-regulated oncolytic adenovirus vector that can replicate and induce a cytopathic effect in alpha-fetoprotein (AFP)-positive HCC cell lines with p53 dysfunction was successfully constructed. Two therapeutic genes, suppressor of cytokine signaling 3 (SOCS3) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), were chosen and incorporated into this vector system, respectively. The combined treatment of AFP-D55-SOCS3 and AFP-D55-TRAIL (2:3 ratio) exhibited potent antitumor activity in AFP-positive HCC cell lines compared with any other treatment both in vitro and in vivo. Specific replication and low progeny yield in AFP-positive HCC cell lines rendered these double-regulated oncolytic adenoviruses remarkably safe. Our data demonstrated that restoration of SOCS3, which inhibits the JAK/STAT3 pathway, by AFP-D55-SOCS3 not only could antagonize HCC therapeutic resistance to TRAIL and adenoviruses, but could also induce cell cycle arrest in HCC cell lines. SOCS3 could down-regulate Cyclin D1 and anti-apoptotic proteins such as XIAP, Survivin, Bcl-xL, and Mcl-1, which are responsible for the synergistic inhibitory effects of AFP-D55-SOCS3 and AFP-D55-TRAIL. Dual gene and double-regulated oncolytic adenoviruses may provide safety and excellent antitumor effects for liver cancer, which is the advantage of a cancer-targeting gene virotherapy strategy.
Collapse
Affiliation(s)
- Rui-Cheng Wei
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Jiang G, Xin Y, Zheng JN, Liu YQ. Combining conditionally replicating adenovirus-mediated gene therapy with chemotherapy: a novel antitumor approach. Int J Cancer 2011; 129:263-74. [PMID: 21509783 DOI: 10.1002/ijc.25948] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Accepted: 01/05/2011] [Indexed: 12/16/2022]
Abstract
Despite significant improvements in diagnosis and innovations in the therapy of specific cancers, effective treatment of neoplastic diseases still presents major challenges. Recent studies have shown that conditionally replicating adenoviruses (CRAds) not only have the ability to destroy cancer cells but may also be potential vectors for the expression of therapeutic genes. Several studies in animal models have demonstrated that the combination of CRAds-mediated gene therapy and chemotherapy has greater therapeutic benefit than either treatment modality alone. In this review, an overview of specifications for a novel antitumor approach combining CRAd-gene therapy and chemotherapy is provided and recent progress in this field is discussed.
Collapse
Affiliation(s)
- Guan Jiang
- Center for Disease Control and Prevention of Xuzhou City, Xuzhou 221006, China
| | | | | | | |
Collapse
|
36
|
Cao X, Yang M, Wei RC, Zeng Y, Gu JF, Huang WD, Yang DQ, Li HL, Ding M, Wei N, Zhang KJ, Xu B, Liu XR, Qian QJ, Liu XY. Cancer targeting Gene-Viro-Therapy of liver carcinoma by dual-regulated oncolytic adenovirus armed with TRAIL gene. Gene Ther 2011; 18:765-77. [PMID: 21412282 DOI: 10.1038/gt.2011.16] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Liver cancer is a common and aggressive malignancy, but available treatment approaches remain suboptimal. Cancer targeting Gene-Viro-Therapy (CTGVT) has shown excellent anti-tumor effects in a preclinical study. CTGVT takes advantage of both gene therapy and virotherapy by incorporating an anti-tumor gene into an oncolytic virus vector. Potent anti-tumor activity is achieved by virus replication and exogenous expression of the anti-tumor gene. A dual-regulated oncolytic adenoviral vector designated Ad·AFP·E1A·E1B (Δ55) (Ad·AFP·D55 for short thereafter) was constructed by replacing the native viral E1A promoter with the simian virus 40 enhancer/α-fetoprotein (AFP) composite promoter (AFPep) based on an E1B-55K-deleted construct, ZD55. Ad·AFP·D55 showed specific replication and cytotoxicity in AFP-positive hepatoma cells. It also showed enhanced safety in normal cells when compared with the mono-regulated vector ZD55. To improve the anti-hepatoma activities of the virus, the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) gene was introduced into Ad·AFP·D55. Ad·AFP·D55-TRAIL exhibited remarkable anti-tumor activities in vitro and in vivo. Treatment with Ad·AFP·D55-TRAIL can induce both autophagy owing to the Ad·AFP·D55 vector and caspase-dependent apoptosis owing to the TRAIL protein. Therefore, Ad·AFP·D55-TRAIL could be a potential anti-hepatoma agent with anti-tumor activities due to AFP-specific replication and TRAIL-induced apoptosis.
Collapse
Affiliation(s)
- X Cao
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Lu Y, Madu CO. Viral-based gene delivery and regulated gene expression for targeted cancer therapy. Expert Opin Drug Deliv 2010; 7:19-35. [PMID: 19947888 DOI: 10.1517/17425240903419608] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
IMPORTANCE OF THE FIELD Cancer is both a major health concern and a care-cost issue in the US and the rest of the world. It is estimated that there will be a total of 1,479,350 new cancer cases and 562,340 cancer deaths in 2009 within the US alone. One of the major obstacles in cancer therapy is the ability to target specifically cancer cells. Most existing chemotherapies and other routine therapies (such as radiation therapy and hormonal manipulation) use indiscriminate approaches in which both cancer cells and non-cancerous surrounding cells are treated equally by the toxic treatment. As a result, either the cancer cell escapes the toxic dosage necessary for cell death and consequently resumes replication, or an adequate lethal dose that kills the cancer cell also causes the cancer patient to perish. Owing to this dilemma, cancer- or organ/tissue-specific targeting is greatly desired for effective cancer treatment and the reduction of side effect cytotoxicity within the patient. AREAS COVERED IN THIS REVIEW In this review, the strategies of targeted cancer therapy are discussed, with an emphasis on viral-based gene delivery and regulated gene expression. WHAT THE READER WILL GAIN Numerous approaches and updates in this field are presented for several common cancer types. TAKE HOME MESSAGE A summary of existing challenges and future directions is also included.
Collapse
Affiliation(s)
- Yi Lu
- University of Tennessee Health Science Center, Department of Pathology and Laboratory Medicine, Cancer Research Building, Room 218, 19 South Manassas Street, Memphis, TN 38163, USA.
| | | |
Collapse
|
38
|
Dichloroacetate (DCA) enhances tumor cell death in combination with oncolytic adenovirus armed with MDA-7/IL-24. Mol Cell Biochem 2010; 340:31-40. [PMID: 20165905 DOI: 10.1007/s11010-010-0397-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Accepted: 02/03/2010] [Indexed: 11/24/2022]
|
39
|
Xiao T, Fan JK, Huang HL, Gu JF, Li LY, Liu XY. VEGI-armed oncolytic adenovirus inhibits tumor neovascularization and directly induces mitochondria-mediated cancer cell apoptosis. Cell Res 2009; 20:367-78. [DOI: 10.1038/cr.2009.126] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
40
|
Zhang Z, Huang Y, Newman K, Gu J, Zhang X, Wu H, Zhao M, Xianyu Z, Liu X. Reexpression of human somatostatin receptor gene 2 gene mediated by oncolytic adenovirus increases antitumor activity of tumor necrosis factor-related apoptosis-inducing ligand against pancreatic cancer. Clin Cancer Res 2009; 15:5154-60. [PMID: 19671855 DOI: 10.1158/1078-0432.ccr-09-0025] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
PURPOSE Pancreatic cancer continues to pose an enormous challenge to clinicians and cancer scientists. Clinical studies show that tumor necrosis factor-related apoptosis- inducing ligand (TRAIL) exerts a potent and tumor-specific proapoptotic activity. However, most pancreatic cancer cells are resistant to TRAIL therapy. Human somatostatin receptor gene 2 (hSSTr2) is lost in 90% of pancreatic carcinoma. Oncolytic viruses are able to selectively lyse cancer cells and represent a promising novel anticancer therapy. Here, we investigated whether oncolytic adenovirus-mediated reexpression of hSSTr2 would enhance TRAIL-induced antitumor efficacy against pancreatic cancer. EXPERIMENTAL DESIGN The antitumor efficacies of combined or single treatment of hSSTr2 and TRAIL mediated by oncolytic adenovirus were compared in pancreatic cancer cell culture and xenografts. The mechanisms involved in hSSTr2-induced sensitization to TRAIL were studied. RESULTS Oncolytic adenovirus-mediated reexpression of hSSTr2 potentiated TRAIL-induced tumor growth inhibition in vitro and in vivo. Reexpression of hSSTr2 augmented TRAIL-induced apoptosis against pancreatic cancer cells via up-regulation of death receptor 4 and down-regulation of Bcl-2. CONCLUSIONS hSSTr2 restoration mediated by oncolytic adenovirus enhances TRAIL-induced antitumor efficacy against pancreatic cancer. Combined treatment with oncolytic adenovirus-mediated hSSTr2 and TRAIL gene provides the rationale for a clinical trial in pancreatic cancer.
Collapse
Affiliation(s)
- Zhenwei Zhang
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Cody JJ, Douglas JT. Armed replicating adenoviruses for cancer virotherapy. Cancer Gene Ther 2009; 16:473-88. [PMID: 19197323 PMCID: PMC2683179 DOI: 10.1038/cgt.2009.3] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2008] [Revised: 09/15/2008] [Accepted: 10/15/2008] [Indexed: 12/22/2022]
Abstract
Conditionally replicating adenoviruses (CRAds) have many advantages as agents for cancer virotherapy and have been safely used in human clinical trials. However, replicating adenoviruses have been limited in their ability to eliminate tumors by oncolysis. Thus, the efficacy of these agents must be improved. To this end, CRAds have been engineered to express therapeutic transgenes that exert antitumor effects independent of direct viral oncolysis. These transgenes can be expressed under native gene control elements, in which case placement within the genome determines the expression profile, or they can be controlled by exogenous promoters. The therapeutic transgenes used to arm replicating adenoviruses can be broadly classified into three groups. There are those that mediate killing of the infected cell, those that modulate the tumor microenvironment and those with immunomodulatory functions. Overall, the studies to date in animal models have shown that arming a CRAd with a rationally chosen therapeutic transgene can improve its antitumor efficacy over that of an unarmed CRAd. However, a number of obstacles must be overcome before the full potential of armed CRAds can be realized in the human clinical context. Hence, strategies are being developed to permit intravenous delivery to disseminated cancer cells, overcome the immune response and enable in vivo monitoring of the biodistribution and activity of armed CRAds.
Collapse
Affiliation(s)
- James J. Cody
- Division of Human Gene Therapy, Departments of Medicine, Obstetrics & Gynecology, Pathology and Surgery, and the Gene Therapy Center, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | - Joanne T. Douglas
- Division of Human Gene Therapy, Departments of Medicine, Obstetrics & Gynecology, Pathology and Surgery, and the Gene Therapy Center, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| |
Collapse
|
42
|
Wu J, Hecker JG, Chiamvimonvat N. Antioxidant enzyme gene transfer for ischemic diseases. Adv Drug Deliv Rev 2009; 61:351-63. [PMID: 19233238 PMCID: PMC2691416 DOI: 10.1016/j.addr.2009.01.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2008] [Accepted: 01/28/2009] [Indexed: 02/07/2023]
Abstract
The balance of redox is pivotal for normal function and integrity of tissues. Ischemic insults occur as results of a variety of conditions, leading to an accumulation of reactive oxygen species (ROS) and an imbalanced redox status in the tissues. The oxidant stress may activate signaling mechanisms provoking more toxic events, and eventually cause tissue damage. Therefore, treatments with antioxidants, free radical scavengers and their mimetics, as well as gene transfer approaches to overexpress antioxidant genes represent potential therapeutic options to correct the redox imbalance. Among them, antioxidant gene transfer may enhance the production of antioxidant scavengers, and has been employed to experimentally prevent or treat ischemic injury in cardiovascular, pulmonary, hepatic, intestinal, central nervous or other systems in animal models. With improvements in vector systems and delivery approaches, innovative antioxidant gene therapy has conferred better outcomes for myocardial infarction, reduced restenosis after coronary angioplasty, improved the quality and function of liver grafts, as well as outcome of intestinal and cerebral ischemic attacks. However, it is crucial to be mindful that like other therapeutic armentarium, the efficacy of antioxidant gene transfer requires extensive preclinical investigation before it can be used in patients, and that it may have unanticipated short- or long-term adverse effects. Thus, it is critical to balance between the therapeutic benefits and potential risks, to develop disease-specific antioxidant gene transfer strategies, to deliver the therapy with an optimal time window and in a safe manner. This review attempts to provide the rationale, the most effective approaches and the potential hurdles of available antioxidant gene transfer approaches for ischemic injury in various organs, as well as the possible directions of future preclinical and clinical investigations of this highly promising therapeutic modality.
Collapse
Affiliation(s)
- Jian Wu
- Department of Internal Medicine, Transplant Research Program, University of California, Davis Medical Center, Sacramento, CA 95817, USA.
| | | | | |
Collapse
|
43
|
Huang XY, Wang L, Huang ZL, Zheng Q, Li QS, Tang ZY. Herbal extract "Songyou Yin" inhibits tumor growth and prolongs survival in nude mice bearing human hepatocellular carcinoma xenograft with high metastatic potential. J Cancer Res Clin Oncol 2009; 135:1245-55. [PMID: 19277711 DOI: 10.1007/s00432-009-0566-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2008] [Accepted: 02/16/2009] [Indexed: 12/20/2022]
Abstract
PURPOSE Chinese herbs have become a focus of interest in cancer treatment. This study evaluates the effect of the herbal compound extract "Songyou Yin" (containing Salvia miltiorrhiza Bge.-danshen and other four herbs) on hepatocellular carcinoma (HCC). METHODS Human HCC cell line MHCC97H with high-metastatic potential was employed for in vitro study. In vivo study was conducted in nude mice bearing HCC orthotopic xenograft with MHCC97H. RESULTS In vitro, "Songyou Yin" caused dramatic attenuation of tumor proliferation by induction of apoptosis that was associated with caspase-3 activation, and inhibit invasiveness of MHCC97H via reducing matrix metalloproteinase-2 (MMP2) activity. In vivo, "Songyou Yin" minimized cancer-related body weight loss of mice bearing tumors without distinct toxicity, and inhibited tumor growth with stepwise increased dosage of "Songyou Yin" and accorded with the expression of proliferating cell nuclear antigen. Moreover, "Songyou Yin" inhibited tumor growth was associated with an increased TUNEL-positive apoptosis as well as a decreased microvessel density and vascular endothelial growth factor (VEGF) abundance, and inhibited tumor invasion via down-regulation of MMP2. The lung metastatic extent was decreased (p < 0.01, compared with control). The life span of nude mice bearing xenografts was 75.0 +/- 3.9 days in "Songyou Yin" group, whereas it was 52.0 +/- 2.3 days in the control (p < 0.001). CONCLUSIONS Nontoxic herbal compound extract "Songyou Yin" inhibited tumor growth and prolonged survival, via inducing apoptosis and down-regulation of MMP2 and VEGF, which indicated its potential use in patients with advanced HCC.
Collapse
Affiliation(s)
- Xiu-Yan Huang
- Department of General Surgery, The 6th People's Hospital Affiliated to Shanghai Jiaotong University, Shanghai 200233, China
| | | | | | | | | | | |
Collapse
|
44
|
He LF, Gu JF, Tang WH, Fan JK, Wei N, Zou WG, Zhang YH, Zhao LL, Liu XY. Significant antitumor activity of oncolytic adenovirus expressing human interferon-beta for hepatocellular carcinoma. J Gene Med 2009; 10:983-92. [PMID: 18618506 DOI: 10.1002/jgm.1231] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Human interferon-beta (IFN-beta) has been widely used in gene therapy for its antitumor activity but its therapeutic effect is limited. The conditionally replicative adenovirus ONYX-015 (a E1B-55-kDa-deleted adenovirus) targets well to tumor cells, but is not potent enough to cause significant tumor regression. To solve these problems, a tumor-selective replicating adenovirus expressing IFN-beta was constructed in this study. METHODS The oncolytic adenoviruses were generated by homologous recombination in packaging cells. The expression of the IFN-beta protein was detected by enzyme-linked immunosorbent assay (ELISA). The antitumor efficacy of ZD55-IFN-beta was evaluated in cell lines and human hepatocellular carcinoma xenografts in nude mice. RESULTS ZD55-IFN-beta can express much more IFN-beta than Ad-IFN-beta because of the replication of the ZD55 vector. Our data showed that ZD55-IFN-beta could exert a strong cytopathic effect on tumor cells (about 100-fold higher than Ad-IFN-beta or ONYX-015). Moreover, no obvious cytotoxic or apoptotic effects were detected in normal cells infected with ZD55-IFN-beta. CONCLUSIONS The antitumor efficacy of IFN-beta could be significantly improved due to the increased gene expression level from the tumor-selective replicating vector. The oncolytic adenovirus expressing IFN-beta may provide a novel approach for cancer gene therapy.
Collapse
Affiliation(s)
- Ling Feng He
- Laboratory of Cancer Therapy, Institute of Biochemistry and Cell Biology, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Wang Y, Huang F, Cai H, Zhong S, Liu X, Tan WS. Potent antitumor effect of TRAIL mediated by a novel adeno-associated viral vector targeting to telomerase activity for human hepatocellular carcinoma. J Gene Med 2008; 10:518-26. [PMID: 18338833 DOI: 10.1002/jgm.1177] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Adeno-associated virus (AAV) has rapidly become a promising gene delivery vehicle for its excellent advantages of low pathogenicity and long-term gene expression. However, lack of tissue specificity caused low efficiency of AAV transfer to target cells. The promoter of human telomerase reverse transcriptase (hTERT) has been implicated in mediating gene expression in cancer cells as hTERT is transcriptionally upregulated in most cancer cells. Thereby, the hTERT promoter becomes a good candidate to enhance the targeting efficiency of AAV in cancer cells. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) functions as a soluble cytokine to selectively kill various cancer cells without toxicity to most normal cells. It remains to be determined whether the hTERT promoter can efficiently mediate TRAIL gene therapy in cancer cells using AAV vector. METHODS A novel AAV vector containing the TRAIL gene under the control of the hTERT promoter (AAV-hTERT-TRAIL) was generated. The specific expression of hTERT-controlled genes was evaluated in cell lines. The antitumor efficacy of AAV-hTERT-TRAIL was assessed in tumor cell lines and human hepatocellular carcinoma xenograft mouse model. RESULTS TRAIL expression was observed in tumor cells infected with AAV-hTERT-TRAIL at both the protein and mRNA level. AAV-hTERT-TRAIL displayed cancer-specific cytotoxicity and induced tumor cell apoptosis. Moreover, in animal experiments, intratumoral administration of AAV-hTERT-TRAIL significantly suppressed the growth of xenograft tumors and resulted in tumor cell death. CONCLUSIONS AAVs in combination with hTERT-mediated therapeutic gene expression provide a promising targeting approach for developing effective therapy for human cancers. These data suggest that AAV-hTERT-TRAIL is a potent therapeutic agent for cancer therapy.
Collapse
Affiliation(s)
- Yigang Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | | | | | | | | | | |
Collapse
|
46
|
Inhibition of renal cancer cell growth in vitro and in vivo with oncolytic adenovirus armed short hairpin RNA targeting Ki-67 encoding mRNA. Cancer Gene Ther 2008; 16:20-32. [PMID: 18690204 DOI: 10.1038/cgt.2008.61] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
RNA interference (RNAi) has been proved to be a powerful tool for gene knockdown purpose and holds great promise for the treatment of cancer. Our previous study demonstrated that the reduction of Ki-67 expression by means of chemically synthesized siRNAs and shRNAs expressed from plasmid resulted in proliferation inhibition in human renal carcinoma cells. In this study, we constructed a novel oncolytic adenovirus-based shRNA expression system, ZD55-Ki67, and explored ZD55-Ki67-mediated RNAi for Ki-67 gene silencing. Our results showed that ZD55-Ki67 could induce silencing of the Ki-67 gene effectively, allow for efficient tumor-specific viral replication and induce the apoptosis of tumor cells effectively in vitro and in nude mice. We conclude that combining shRNA gene therapy and oncolytic virotherapy can enhance antitumor efficacy as a result of synergism between CRAd oncolysis and shRNA antitumor responses.
Collapse
|
47
|
Adenovirus-mediated HCCS1 overexpression elicits a potent antitumor efficacy on human colorectal cancer and hepatoma cells both in vitro and in vivo. Cancer Gene Ther 2008; 15:808-16. [DOI: 10.1038/cgt.2008.52] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
48
|
Shashkova EV, Kuppuswamy MN, Wold WSM, Doronin K. Anticancer activity of oncolytic adenovirus vector armed with IFN-alpha and ADP is enhanced by pharmacologically controlled expression of TRAIL. Cancer Gene Ther 2007; 15:61-72. [PMID: 17992200 DOI: 10.1038/sj.cgt.7701107] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We have previously described oncolytic adenovirus (Ad) vectors KD3 and KD3-interferon (IFN) that were rendered cancer-specific by mutations in the E1A region of Ad; these mutations abolish binding of E1A proteins to p300/CBP and pRB. The antitumor activity of the vectors was enhanced by overexpression of the Adenovirus Death Protein (ADP, E3-11.6K) and by replication-linked expression of IFN-alpha. We hypothesized that the anticancer efficacy of the KD3-IFN vector could be further improved by expression of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). E1-deleted Ad vectors were constructed carrying reporter genes for enhanced green fluorescent protein or secreted placental alkaline phosphatase (SEAP) and a therapeutic gene for TRAIL under control of the TetON system. Expression of the genes was increased in the presence of a helper virus and the inducer doxycycline such that up to 231-fold activation of expression for the TetON-SEAP vector was obtained. Coinfection with TetON-TRAIL augmented oncolytic activity of KD3 and KD3-IFN in vitro. Induction of TRAIL expression did not reduce the yield of progeny virus. Combination of TetON-TRAIL and KD3-IFN produced superior antitumor activity in vivo as compared with either vector alone demonstrating the efficacy of a four-pronged cancer gene therapy approach, which includes Ad oncolysis, ADP overexpression, IFN-alpha-mediated immunotherapy, and pharmacologically controlled TRAIL activity.
Collapse
|
49
|
Zhang Y, Venugopal SK, He S, Liu P, Wu J, Zern MA. Ethanol induces apoptosis in hepatocytes by a pathway involving novel protein kinase C isoforms. Cell Signal 2007; 19:2339-50. [PMID: 17728104 DOI: 10.1016/j.cellsig.2007.07.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2007] [Revised: 07/11/2007] [Accepted: 07/18/2007] [Indexed: 11/29/2022]
Abstract
UNLABELLED Ethanol abuse is one of the major etiologies of cirrhosis. Ethanol has been shown to induce apoptosis via activation of oxidative stress, mitogen-activated protein kinases (MAPK), and tyrosine kinases. However, there is a paucity of data that examine the interplay among these molecules. In the present study we have systematically elucidated the role of novel protein kinase C isoforms (nPKC; PKCdelta and PKCepsilon) in ethanol-induced apoptosis in hepatocytes. Ethanol enhanced membrane translocation of PKCdelta and PKCepsilon, which was associated with the phosphorylation of p38MAPK, p42/44MAPK and JNK1/2, and the nuclear translocation of NF-kappaB and AP-1. This resulted in increased apoptosis in primary rat hepatocytes. Inhibition of both PKCdelta and PKCepsilon resulted in a decreased MAPK activation, decreased nuclear translocation of NF-kappaB and AP-1, and inhibition of apoptosis. In addition, ethanol activated the tyrosine phosphorylation of PKCdelta via tyrosine kinase in hepatocytes. The tyrosine phosphorylated PKCdelta was cleaved by caspase-3 and these fragments were translocated to the nucleus. Inhibition of ethanol-induced oxidative stress blocked the membrane translocation of PKCdelta and PKCepsilon, and the tyrosine phosphorylation of PKCdelta in hepatocytes. Inhibition of oxidative stress, tyrosine kinase or caspase-3 activity caused a decreased nuclear translocation of PKCdelta in response to ethanol, and was associated with less apoptosis. CONCLUSION These results provide a newly-described mechanism by which ethanol induces apoptosis via activation of nPKC isoforms in hepatocytes.
Collapse
Affiliation(s)
- Yanhong Zhang
- Department of Internal Medicine, Transplant Research Program, University of California, Davis Medical Center, Sacramento, CA 95817, USA
| | | | | | | | | | | |
Collapse
|
50
|
White SJ, Kasman LM, Kelly MM, Lu P, Spruill L, McDermott PJ, Voelkel-Johnson C. Doxorubicin generates a proapoptotic phenotype by phosphorylation of elongation factor 2. Free Radic Biol Med 2007; 43:1313-21. [PMID: 17893044 PMCID: PMC2084083 DOI: 10.1016/j.freeradbiomed.2007.06.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2007] [Revised: 06/21/2007] [Accepted: 06/23/2007] [Indexed: 11/24/2022]
Abstract
We have previously shown that doxorubicin sensitizes prostate cancer cells to tumor-necrosis-factor-related apoptosis-inducing ligand (TRAIL). Sensitization correlated with decreased expression of the antiapoptotic cellular FLICE-like inhibitor protein (cFLIP(S)). The decrease in cFLIP(S) could not be explained by transcriptional regulation or increased degradation, leading us to focus on translational mechanisms. In this study, we found that doxorubicin caused strong and sustained phosphorylation of elongation factor 2 (EF-2), which interferes with protein elongation. Phosphorylation of EF-2 appeared to occur in a kinase-independent manner. Treatment with hydrogen peroxide recapitulated the events observed after doxorubicin treatment. In addition, cells treated with hydrogen peroxide expressed less X-linked inhibitor of apoptosis protein (XIAP) and survivin which, like cFLIP(S), are short-half-life proteins with an antiapoptotic function while expression levels of DR5, caspases-8, -9, -3, and Bax are maintained. The doxorubicin-mediated decrease in cFLIP(S) and XIAP and the TRAIL-induced apoptosis were prevented by pretreatment with an iron chelator, indicating that expression of these proteins was affected by free radical generation upon interaction of iron with doxorubicin. In conclusion, our data suggest that free radicals can affect the phosphorylation of EF-2 resulting in a net loss of short-half-life proteins such as cFLIP(S) and XIAP, leaving a cell more vulnerable to apoptotic stimuli.
Collapse
Affiliation(s)
- Shai J. White
- Department of Microbiology & Immunology, Medical University of South Carolina, 173 Ashley Ave Charleston SC 29425, USA
| | - Laura M. Kasman
- Department of Microbiology & Immunology, Medical University of South Carolina, 173 Ashley Ave Charleston SC 29425, USA
| | - Margaret M. Kelly
- Hollings Cancer Center, Medical University of South Carolina, 173 Ashley Ave Charleston SC 29425, USA
| | - Ping Lu
- Department of Microbiology & Immunology, Medical University of South Carolina, 173 Ashley Ave Charleston SC 29425, USA
| | - Laura Spruill
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, 173 Ashley Ave Charleston SC 29425, USA
| | - Paul J. McDermott
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, 173 Ashley Ave Charleston SC 29425, USA
| | - Christina Voelkel-Johnson
- Department of Microbiology & Immunology, Medical University of South Carolina, 173 Ashley Ave Charleston SC 29425, USA
| |
Collapse
|